natriuretic-peptide--c-type has been researched along with Disease-Models--Animal* in 37 studies
2 review(s) available for natriuretic-peptide--c-type and Disease-Models--Animal
Article | Year |
---|---|
[Genetic basis for skeletal disease. CNP therapy for achondroplasia].
By using transgenic and knockout mice, we have elucidated that C-type natriuretic peptide (CNP) is a potent stimulator of endochondral bone growth. Furthermore, loss-of-function mutations in the gene coding for guanylyl cyclase-B (GC-B) , the specific receptor for CNP, have been proved to cause impaired skeletal growth in humans. Following these results, we have started to translate the stimulatory effect of CNP on endochondral bone growth into the therapy for patients with achondroplasia, and have shown that targeted overexpression of CNP in cartilage or systemic administration of CNP reverses the impaired skeletal growth of mice model of achondroplasia. Topics: Achondroplasia; Animals; Cartilage; Disease Models, Animal; Humans; Mice; Mice, Knockout; Mice, Transgenic; Mutation; Natriuretic Peptide, C-Type; Receptors, Atrial Natriuretic Factor | 2010 |
The role of the natriuretic peptides in the cardiovascular system.
The discovery of the natriuretic peptide family was a breakthrough in modern cardiovascular physiology as it provided a direct link between the heart and the kidneys in the regulation of natriuresis. Along with vasopressin and the renin-angiotensin-aldosterone system, the natriuretic peptides comprise the key peptides on which our present understanding of neuroendocrine regulation of the cardiovascular system is based. Three natriuretic peptides have been identified; the A-type, B-type and C-type natriuretic peptides. The former two, the A- and B-type natriuretic peptides, function mainly in the cardiovascular system and comprise the cardiac natriuretic peptides. Together with our increased understanding of the neurohormonal regulation of the cardiovascular system in recent years, the discovery of the natriuretic peptide family was important in the establishment of the new field of cardiovascular endocrinology. Topics: Animals; Atrial Natriuretic Factor; Cardiovascular Physiological Phenomena; Disease Models, Animal; Humans; Natriuresis; Natriuretic Peptide, Brain; Natriuretic Peptide, C-Type; Peptides | 2001 |
35 other study(ies) available for natriuretic-peptide--c-type and Disease-Models--Animal
Article | Year |
---|---|
Atrial Fibrillation in Aging and Frail Mice: Modulation by Natriuretic Peptide Receptor C.
[Figure: see text]. Topics: Aging; Animals; Atrial Fibrillation; Biomarkers; Disease Models, Animal; Female; Heart Atria; Male; Mice; Natriuretic Peptide, C-Type | 2021 |
Stabilization of Perivascular Mast Cells by Endothelial CNP (C-Type Natriuretic Peptide).
Activated perivascular mast cells (MCs) participate in different cardiovascular diseases. Many factors provoking MC degranulation have been described, while physiological counterregulators are barely known. Endothelial CNP (C-type natriuretic peptide) participates in the maintenance of vascular barrier integrity, but the target cells and mechanisms are unclear. Here, we studied whether MCs are regulated by CNP. Approach and Results: In cultured human and murine MCs, CNP activated its specific GC (guanylyl cyclase)-B receptor and cyclic GMP signaling. This enhanced cyclic GMP-dependent phosphorylation of the cytoskeleton-associated VASP (vasodilator-stimulated phosphoprotein) and inhibited ATP-evoked degranulation. To elucidate the relevance in vivo, mice with a floxed GC-B (. CNP, via GC-B/cyclic GMP signaling, stabilizes resident perivascular MCs at baseline and prevents their excessive activation under pathological conditions. Thereby CNP contributes to the maintenance of vascular integrity in physiology and disease. Topics: Adenosine Triphosphate; Animals; Capillary Permeability; Cell Adhesion Molecules; Cell Degranulation; Cell Line; Cyclic GMP; Disease Models, Animal; Endothelial Cells; Mast Cells; Mice, 129 Strain; Mice, Inbred C57BL; Mice, Knockout; Microfilament Proteins; Myocardial Reperfusion Injury; Natriuretic Peptide, C-Type; Neutrophil Infiltration; Paracrine Communication; Phosphoproteins; Phosphorylation; Receptors, Atrial Natriuretic Factor; Signal Transduction; Thrombosis | 2020 |
Pharmacokinetics and intraocular pressure-lowering activity of TAK-639, a novel C-type natriuretic peptide analog, in rabbit, dog, and monkey.
Topics: Administration, Topical; Animals; Antihypertensive Agents; Aqueous Humor; Chromatography, Liquid; Disease Models, Animal; Dogs; Dose-Response Relationship, Drug; Female; Glaucoma, Open-Angle; Intraocular Pressure; Macaca fascicularis; Natriuretic Peptide, C-Type; Rabbits | 2019 |
C-type natriuretic peptide improves growth retardation in a mouse model of cardio-facio-cutaneous syndrome.
Cardio-facio-cutaneous (CFC) syndrome, a genetic disorder caused by germline mutations in BRAF, KRAS, MAP2K1 and MAP2K2, is characterized by growth retardation, heart defects, dysmorphic facial appearance and dermatologic abnormalities. We have previously reported that knock-in mice expressing the CFC syndrome-associated mutation, Braf Q241R, showed growth retardation because of gastrointestinal dysfunction. However, other factors associated with growth retardation, including chondrogenesis and endocrinological profile, have not been examined. Here, we show that 3- and 4-week-old BrafQ241R/+ mice have decreased body weight and length, as well as reduced growth plate width in the proximal tibiae. Furthermore, proliferative and hypertrophic chondrocyte zones of the growth plate were reduced in BrafQ241R/+ mice compared with Braf+/+ mice. Immunohistological analysis revealed that extracellular signal-regulated kinase (ERK) activation was enhanced in hypertrophic chondrocytes in BrafQ241R/+ mice. In accordance with growth retardation and reduced growth plate width, decreased serum levels of insulin-like growth factor 1 (IGF-1) and IGF binding protein 3 (IGFBP-3) were observed in BrafQ241R/+ mice at 3 and 4 weeks of age. Treatment with C-type natriuretic peptide (CNP), a stimulator of endochondral bone growth and a potent inhibitor of the FGFR3-RAF1-MEK/ERK signaling, increased body and tail lengths in Braf+/+ and BrafQ241R/+ mice. In conclusion, ERK activation in chondrocytes and low serum IGF-1/IGFBP-3 levels could be associated with the growth retardation observed in BrafQ241R/+ mice. Our data also suggest that CNP is a potential therapeutic target in CFC syndrome. Topics: Animals; Chondrocytes; Disease Models, Animal; Ectodermal Dysplasia; Facies; Failure to Thrive; Germ-Line Mutation; Growth Disorders; Heart Defects, Congenital; Humans; Insulin-Like Growth Factor I; MAP Kinase Kinase 1; MAP Kinase Kinase 2; MAP Kinase Signaling System; Mice; Mice, Inbred ICR; Mutation; Natriuretic Peptide, C-Type; Proto-Oncogene Proteins B-raf | 2019 |
Exogenous C-type natriuretic peptide therapy for impaired skeletal growth in a murine model of glucocorticoid treatment.
Growth retardation is an important side effect of glucocorticoid (GC)-based drugs, which are widely used in various preparations to treat many pediatric diseases. We investigated the therapeutic effect of exogenous CNP-53, a stable molecular form of intrinsic CNP, on a mouse model of GC-induced growth retardation. We found that CNP-53 successfully restored GC-induced growth retardation when both dexamethasone (DEX) and CNP-53 were injected from 4 to 8 weeks old. Notably, CNP-53 was not effective during the first week. From 4 to 5 weeks old, neither CNP-53 in advance of DEX, nor high-dose CNP-53 improved the effect of CNP. Conversely, when CNP-53 was started at 5 weeks old, final body length at 8 weeks old was comparable to that when CNP-53 was started at 4 weeks old. As for the mechanism of resistance to the CNP effect, DEX did not impair the production of cGMP induced by CNP. CNP reduced Erk phosphorylation even under treatment with DEX, while CNP did not changed that of p38 or GSK3β. Collectively, the effect of CNP-53 on GC-induced growth retardation is dependent on age in a mouse model, suggesting adequate and deliberate use of CNP would be effective for GC-induced growth retardation in clinical settings. Topics: Animals; Dexamethasone; Disease Models, Animal; Glucocorticoids; Glycogen Synthase Kinase 3 beta; Growth Disorders; Humans; MAP Kinase Signaling System; Mice; Natriuretic Peptide, C-Type; p38 Mitogen-Activated Protein Kinases | 2019 |
C-type natriuretic peptide attenuates renal osteodystrophy through inhibition of FGF-23/MAPK signaling.
Renal osteodystrophy (ROD) occurs as early as chronic kidney disease (CKD) stage 2 and seems ubiquitous in almost all pediatric patients with CKD stage 5. Fibroblast growth factor (FGF)-23, a bone-derived endocrine regulator of phosphate homeostasis, is overexpressed in CKD and disturbs osteoblast differentiation and matrix mineralization. In contrast, C-type natriuretic peptide (CNP) acts as a potent positive regulator of bone growth. In the present study, we infused CNP into uremic rats and observed whether CNP could attenuate ROD through the inhibition of FGF-23 cascades. In uremic rats, CNP administration significantly alleviated renal dysfunction, calcium phosphate metabolic disorders, hypovitaminosis D, secondary hyperparathyroidism, the decrease in bone turnover markers and retarded bone pathological progression. More importantly, within FGF-23/mitogen-activated protein kinase (MAPK) signaling, the fibroblast growth factor receptor-1, Klotho and alternative (STAT-1/phospho-STAT-1) elements were upregulated by CNP, whereas FGF-23, RAF-1/phospho-RAF-1, and downstream (ERK/phospho-ERK and P38/phospho-P38) elements were paradoxically underexpressed in bone tissue. Therefore, CNP exerts a therapeutic effect on ROD through inhibition of FGF-23/MAPK signaling at the RAF-1 level. Topics: Animals; Bone and Bones; Bone Remodeling; Calcium; Cell Differentiation; Chronic Kidney Disease-Mineral and Bone Disorder; Disease Models, Animal; Fibroblast Growth Factor-23; Fibroblast Growth Factors; Gene Expression Regulation; Humans; Kidney; Male; MAP Kinase Signaling System; Natriuretic Peptide, C-Type; Proto-Oncogene Proteins c-raf; Rats; Rats, Sprague-Dawley; Up-Regulation; Uremia | 2019 |
High level of C-type natriuretic peptide induced by hyperandrogen-mediated anovulation in polycystic ovary syndrome mice.
Topics: Adult; Androgen Receptor Antagonists; Animals; Anovulation; Case-Control Studies; Chorionic Gonadotropin; Disease Models, Animal; Estrogen Receptor Antagonists; Female; HEK293 Cells; Humans; Hyperandrogenism; Mice, Inbred BALB C; Natriuretic Peptide, C-Type; Ovary; Polycystic Ovary Syndrome; Receptors, Androgen; Receptors, Atrial Natriuretic Factor; Receptors, Estrogen; Young Adult | 2018 |
C-type natriuretic peptide analog treatment of craniosynostosis in a Crouzon syndrome mouse model.
Activating mutations of fibroblast growth factor receptors (FGFRs) are a major cause of skeletal dysplasias, and thus they are potential targets for pharmaceutical intervention. BMN 111, a C-type natriuretic peptide analog, inhibits FGFR signaling at the level of the RAF1 kinase through natriuretic peptide receptor 2 (NPR2) and has been shown to lengthen the long bones and improve skull morphology in the Fgfr3Y367C/+ thanatophoric dysplasia mouse model. Here we report the effects of BMN 111 in treating craniosynostosis and aberrant skull morphology in the Fgfr2cC342Y/+ Crouzon syndrome mouse model. We first demonstrated that NPR2 is expressed in the murine coronal suture and spheno-occipital synchondrosis in the newborn period. We then gave Fgfr2cC342Y/+ and Fgfr2c+/+ (WT) mice once-daily injections of either vehicle or reported therapeutic levels of BMN 111 between post-natal days 3 and 31. Changes in skeletal morphology, including suture patency, skull dimensions, and long bone length, were assessed by micro-computed tomography. Although BMN 111 treatment significantly increased long bone growth in both WT and mutant mice, skull dimensions and suture patency generally were not significantly affected. A small but significant increase in the relative length of the anterior cranial base was observed. Our results indicate that the differential effects of BMN 111 in treating various skeletal dysplasias may depend on the process of bone formation targeted (endochondral or intramembranous), the specific FGFR mutated, and/or the specific signaling pathway changes due to a given mutation. Topics: Animals; Animals, Newborn; Craniofacial Dysostosis; Craniosynostoses; Disease Models, Animal; Female; Male; Mice; Mice, Transgenic; Natriuretic Peptide, C-Type; Receptor, Fibroblast Growth Factor, Type 2; Treatment Outcome | 2018 |
Dephosphorylation is the mechanism of fibroblast growth factor inhibition of guanylyl cyclase-B.
Activating mutations in fibroblast growth factor receptor 3 (FGFR3) and inactivating mutations of guanylyl cyclase-B (GC-B, also called NPRB or NPR2) cause dwarfism. FGF exposure inhibits GC-B activity in a chondrocyte cell line, but the mechanism of the inactivation is not known. Here, we report that FGF exposure causes dephosphorylation of GC-B in rat chondrosarcoma cells, which correlates with a rapid, potent and reversible inhibition of C-type natriuretic peptide-dependent activation of GC-B. Cells expressing a phosphomimetic mutant of GC-B that cannot be inactivated by dephosphorylation because it contains glutamate substitutions for all known phosphorylation sites showed no decrease in GC-B activity in response to FGF. We conclude that FGF rapidly inactivates GC-B by a reversible dephosphorylation mechanism, which may contribute to the signaling network by which activated FGFR3 causes dwarfism. Topics: Animals; Chondrocytes; Cyclic GMP; Disease Models, Animal; Dwarfism; Glutamic Acid; Humans; Natriuretic Peptide, C-Type; Phosphorylation; Rats; Receptor, Fibroblast Growth Factor, Type 3; Receptors, Atrial Natriuretic Factor; Signal Transduction | 2017 |
Elevated C-type natriuretic peptide elicits exercise preconditioning-induced cardioprotection against myocardial injury probably via the up-regulation of NPR-B.
To evaluate exercise preconditioning (EP)-induced cardioprotective effects against exercise-induced acute myocardial injury and investigate the alterations of C-type natriuretic peptide (CNP) and its specific receptor, natriuretic peptide receptor B (NPR-B), during EP-induced cardioprotection. Rats were subjected to treadmill exercise as an EP model (4 periods of 10 min each at 30 m/min with intervening periods of rest lasting 10 min). High-intensity exercise was performed 0.5 and 24 h after the EP. EP attenuated high-intensity exercise-induced myocardial injury in both the early and late phases. After EP and high-intensity exercise, CNP and NPR-B levels increased robustly, but no alterations in the plasma CNP were observed. The enhanced NPR-B, plasma and tissue CNP, and its mRNA levels after high-intensity exercise were significantly elevated by EP. These results suggest that cardiac CNP and NPR-B play an important role in EP-mediated cardioprotection against high-intensity exercise-induced myocardial injury in rats. Topics: Animals; Disease Models, Animal; Male; Myocardial Infarction; Myocardium; Natriuretic Peptide, C-Type; Physical Conditioning, Animal; Physical Exertion; Rats, Sprague-Dawley; Receptors, Atrial Natriuretic Factor; Running; Signal Transduction; Time Factors; Up-Regulation | 2017 |
C-type natriuretic peptide prevents kidney injury and attenuates oxidative and inflammatory responses in hemorrhagic shock.
Oxidative stress induced by hemorrhagic shock (HS) initiates a systemic inflammatory response, which leads to subsequent kidney injury. This study assessed the efficacy of c-type natriuretic peptide (CNP) in attenuating kidney injury in a rat model of hemorrhagic shock and resuscitation (HS/R). Sodium pentobarbital-anesthetized adult male Wistar rats underwent HS induced by the withdrawal of blood to a mean arterial pressure of 30-35 mmHg for 50 min. Then, the animals received CNP (25 μg/kg) or vehicle (saline) intravenously, followed byresuscitation with 1.5 times the shed blood volume in the form of normal saline. Mean arterial pressure was measured throughout the experiment, and acid-base status, oxidative stress, inflammation, tissue injury and kidney function were evaluated after resuscitation. CNP infusion reduced the malondialdehyde content, lowered the myeloperoxidase activity and decreased the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β in the kidney. The histologic injury score and the plasma creatinine concentration were also significantly decreased after CNP treatment compared to the vehicle group. CNP treatment ameliorates oxidative stress, the inflammatory response, and consequently acute kidney injury after HS/R. Thus, CNP may represent a promising strategy to improve resuscitation for the treatment of HS and deserves further investigation. Topics: Acute Kidney Injury; Animals; Blood Gas Analysis; Cytokines; Disease Models, Animal; Inflammation; Kidney; Lipid Peroxidation; Male; Natriuretic Peptide, C-Type; Neutrophils; Oxidative Stress; Rats, Wistar; Resuscitation; Shock, Hemorrhagic | 2017 |
Endothelium-Derived C-Type Natriuretic Peptide Contributes to Blood Pressure Regulation by Maintaining Endothelial Integrity.
We previously reported the secretion of C-type natriuretic peptide (CNP) from vascular endothelial cells and proposed the existence of a vascular natriuretic peptide system composed of endothelial CNP and smooth muscle guanylyl cyclase-B (GC-B), the CNP receptor, and involved in the regulation of vascular tone, remodeling, and regeneration. In this study, we assessed the functional significance of this system in the regulation of blood pressure in vivo using vascular endothelial cell-specific CNP knockout and vascular smooth muscle cell-specific GC-B knockout mice. These mice showed neither the skeletal abnormality nor the early mortality observed in systemic CNP or GC-B knockout mice. Endothelial cell-specific CNP knockout mice exhibited significantly increased blood pressures and an enhanced acute hypertensive response to nitric oxide synthetase inhibition. Acetylcholine-induced, endothelium-dependent vasorelaxation was impaired in rings of mesenteric artery isolated from endothelial cell-specific CNP knockout mice. In addition, endothelin-1 gene expression was enhanced in pulmonary vascular endothelial cells from endothelial cell-specific CNP knockout mice, which also showed significantly higher plasma endothelin-1 concentrations and a greater reduction in blood pressure in response to an endothelin receptor antagonist than their control littermates. By contrast, vascular smooth muscle cell-specific GC-B knockout mice exhibited blood pressures similar to control mice, and acetylcholine-induced vasorelaxation was preserved in their isolated mesenteric arteries. Nonetheless, CNP-induced acute vasorelaxation was nearly completely abolished in mesenteric arteries from vascular smooth muscle cell-specific GC-B knockout mice. These results demonstrate that endothelium-derived CNP contributes to the chronic regulation of vascular tone and systemic blood pressure by maintaining endothelial function independently of vascular smooth muscle GC-B. Topics: Animals; Blood Pressure; Disease Models, Animal; Endothelium, Vascular; Hypertension; Mice; Mice, Inbred C57BL; Mice, Knockout; Muscle, Smooth, Vascular; Natriuretic Peptide, C-Type; Vasoconstriction; Vasodilation | 2017 |
C-type natriuretic peptide restores impaired skeletal growth in a murine model of glucocorticoid-induced growth retardation.
Glucocorticoids are widely used for treating autoimmune conditions or inflammatory disorders. Long-term use of glucocorticoids causes impaired skeletal growth, a serious side effect when they are used in children. We have previously demonstrated that C-type natriuretic peptide (CNP) is a potent stimulator of endochondral bone growth. In this study, we investigated the effect of CNP on impaired bone growth caused by glucocorticoids by using a transgenic mouse model with an increased circulating CNP level. Daily administration of a high dose of dexamethasone (DEX) to 4-week-old male wild-type mice for 4weeks significantly shortened their naso-anal length, which was restored completely in DEX-treated CNP transgenic mice. Impaired growth of the long bones and vertebrae by DEX was restored to a large extent in the CNP transgenic background, with recovery in the narrowed growth plate by increased cell volume, whereas the decreased proliferation and increased apoptosis of the growth plate chondrocytes were unaffected. Trabecular bone volume was not changed by DEX treatment, but decreased significantly in a CNP transgenic background. In young male rats, the administration of high doses of DEX greatly decreased N-terminal proCNP concentrations, a marker of CNP production. In organ culture experiments using fetal wild-type murine tibias, longitudinal growth of tibial explants was inhibited by DEX but reversed by CNP. These findings now warrant further study of the therapeutic potency of CNP in glucocorticoid-induced bone growth impairment. Topics: Animals; Bone Development; Disease Models, Animal; Glucocorticoids; Growth Disorders; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Inbred ICR; Mice, Transgenic; Natriuretic Peptide, C-Type; Rats; X-Ray Microtomography | 2016 |
Dipyridamole-induced C-type natriuretic peptide mRNA overexpression in a minipig model of pacing-induced left ventricular dysfunction.
Dipyridamole (DP) restores ischemic tissue blood flow stimulating angiogenesis in eNOS-dependent pathways. C-type natriuretic peptide (CNP) is expected to mimic the migration-stimulatory effect of NO via a cGMP-dependent mechanism. Aim of this study was to assess the role of concomitant treatment with DP on CNP levels in blood and myocardial tissue of minipigs with left ventricular dysfunction (LVD) induced by pacing at 200bpm in the right ventricular apex. Minipigs with DP therapy (DP+, n=4) or placebo (DP-, n=4) and controls (C-SHAM, n=4) underwent 2D-EchoDoppler examination and blood collection before and after 4 weeks of pacing, when cardiac tissue was collected. Histological/immunohistochemical analyses were performed. CNP levels were determined by radioimmunoassay; cardiac CNP, BNP, natriuretic receptors expression by Real-Time PCR. After pacing, cardiac parameters resulted less impaired in DP+ compared to DP-. Histological sections presented normal morphology while the arteriolar density resulted: C-SHAM: 9.0±1.2; DP-: 4.9±0.3; DP+: 6.5±0.6number/mm(2); C-SHAM vs DP- and DP+ p=0.004, p=0.04, respectively. CNP mRNA resulted lower in DP- compared to C-SHAM and DP+ as well as NPR-B (p=0.011, DP- vs DP+). Both NPR-A/NPR-C mRNA expressions were significantly (p<0.001) lower both in DP- and DP+ compared to C-SHAM. BNP mRNA was higher in LVD. CNP plasma levels showed a similar trend with respect to gene expression (C-SHAM: 30.5±15; DP-: 18.6±5.5; DP+: 21.2±4.7pg/ml). These data suggest that DP may serve as a preconditioning agent to increase the protective CNP-mediated endocrine response in LVD. This response, mediated by its specific receptor NPR-B, may offer new insights into molecular targets for treatment of LVD. Topics: Animals; Cardiac Pacing, Artificial; Dipyridamole; Disease Models, Animal; Heart; Natriuretic Peptide, C-Type; Protective Agents; RNA, Messenger; Swine; Swine, Miniature; Up-Regulation; Ventricular Dysfunction, Left | 2015 |
CNP signal pathway up-regulated in rectum of depressed rats and the interventional effect of Xiaoyaosan.
To investigate the distribution and expression of C-type natriuretic peptide (CNP)/natriuretic peptide receptor B (NPR-B) in the rectum of a rodent depression model and the interventional effect of Xiaoyaosan (XYS).. Male rats (n = 45) of clean grade (200 ± 20 g) were divided into five groups after one week of adaptive feeding: primary control, depression model, low dose XYS, middle dose XYS, and high dose XYS. The animal experiment continued for 3 wk. Primary controls were fed normally ad libitum. The rats of all other groups were raised in solitary and exposed to classic chronic mild unpredictable stimulation each day. XYS groups were perfused intragastrically with low dose, middle dose, and high dose XYS one hour before stimulation. Primary control and depression model groups were perfused intragastrically with normal saline under similar conditions as the XYS groups. Three weeks later, all rats were sacrificed, and the expression levels of CNP and NPR-B in rectum tissues were analyzed by immunohistochemistry, real-time polymerase chain reaction, and Western blotting.. CNP and NPR-B were both expressed in the rectum tissues of all rats. However, the expression levels of CNP and NPR-B at both gene and protein levels in the depression model group were significantly higher when compared to the primary control group (n = 9; P < 0.01). XYS intervention markedly inhibited the expression levels of CNP and NPR-B in depressed rats. The expression levels of CNP and NPR-B in the high dose XYS group did not significantly differ from the expression levels in the primary control group. Additionally, the high and middle dose XYS groups (but not the low dose group) significantly exhibited lower CNP and NPR-B expression levels in the rectum tissues of the respectively treated rats compared to the untreated depression model cohort (n = 9; P < 0.01).. The CNP/NPR-B pathway is upregulated in the rectum of depressed rats and may be one mechanism for depression-associated digestive disorders. XYS antagonizes this pathway at least partially. Topics: Animals; Antidepressive Agents; Behavior, Animal; Depression; Disease Models, Animal; Dose-Response Relationship, Drug; Drugs, Chinese Herbal; Male; Natriuretic Peptide, C-Type; Rats, Sprague-Dawley; Receptors, Atrial Natriuretic Factor; Rectum; RNA, Messenger; Signal Transduction; Time Factors; Up-Regulation | 2015 |
Implications for Cardiac Function Following Rescue of the Dystrophic Diaphragm in a Mouse Model of Duchenne Muscular Dystrophy.
Duchenne muscular dystrophy (DMD) is caused by absence of the integral structural protein, dystrophin, which renders muscle fibres susceptible to injury and degeneration. This ultimately results in cardiorespiratory dysfunction, which is the predominant cause of death in DMD patients, and highlights the importance of therapeutic targeting of the cardiorespiratory system. While there is some evidence to suggest that restoring dystrophin in the diaphragm improves both respiratory and cardiac function, the role of the diaphragm is not well understood. Here using exon skipping oligonucleotides we predominantly restored dystrophin in the diaphragm and assessed cardiac function by MRI. This approach reduced diaphragmatic pathophysiology and markedly improved diaphragm function but did not improve cardiac function or pathophysiology, with or without exercise. Interestingly, exercise resulted in a reduction of dystrophin protein and exon skipping in the diaphragm. This suggests that treatment regimens may require modification in more active patients. In conclusion, whilst the diaphragm is an important respiratory muscle, it is likely that dystrophin needs to be restored in other tissues, including multiple accessory respiratory muscles, and of course the heart itself for appropriate therapeutic outcomes. This supports the requirement of a body-wide therapy to treat DMD. Topics: Amino Acid Sequence; Animals; Atrial Natriuretic Factor; Blotting, Western; Diaphragm; Disease Models, Animal; Dystrophin; Gene Expression; Heart; Humans; Magnetic Resonance Imaging; Male; Mice, Inbred C57BL; Mice, Inbred mdx; Morpholinos; Muscle, Skeletal; Muscular Dystrophy, Duchenne; NADPH Oxidase 4; NADPH Oxidases; Natriuretic Peptide, C-Type; Peptides; Physical Conditioning, Animal; Protein Precursors; Radiography; Reverse Transcriptase Polymerase Chain Reaction | 2015 |
Noninvasive and quantitative live imaging reveals a potential stress-responsive enhancer in the failing heart.
Recent advances in genome analysis have enabled the identification of numerous distal enhancers that regulate gene expression in various conditions. However, the enhancers involved in pathological conditions are largely unknown because of the lack of in vivo quantitative assessment of enhancer activity in live animals. Here, we established a noninvasive and quantitative live imaging system for monitoring transcriptional activity and identified a novel stress-responsive enhancer of Nppa and Nppb, the most common markers of heart failure. The enhancer is a 650-bp fragment within 50 kb of the Nppa and Nppb loci. A chromosome conformation capture (3C) assay revealed that this distal enhancer directly interacts with the 5'-flanking regions of Nppa and Nppb. To monitor the enhancer activity in a live heart, we established an imaging system using the firefly luciferase reporter. Using this imaging system, we observed that the novel enhancer activated the reporter gene in pressure overload-induced failing hearts (failing hearts: 5.7±1.3-fold; sham-surgery hearts: 1.0±0.2-fold; P<0.001, repeated-measures ANOVA). This method will be particularly useful for identifying enhancers that function only during pathological conditions. Topics: 5' Flanking Region; Adrenergic alpha-1 Receptor Agonists; Animals; Animals, Newborn; Atrial Natriuretic Factor; Cells, Cultured; Disease Models, Animal; Enhancer Elements, Genetic; Gene Expression Regulation; Heart Failure; Humans; Luciferases; Luminescent Measurements; Mice; Mice, Inbred C57BL; Mice, Transgenic; Natriuretic Peptide, Brain; Natriuretic Peptide, C-Type; Protein Precursors; Rats; Rats, Wistar; Reverse Transcriptase Polymerase Chain Reaction; Stress, Physiological | 2014 |
Heterotaxy-spectrum heart defects in Zic3 hypomorphic mice.
Mutations in Zinc Finger Protein of the Cerebellum 3 (ZIC3) cause X-linked heterotaxy and isolated cardiovascular malformations. Recent data suggest a potential cell-autonomous role for Zic3 in myocardium via regulation of Nppa and Tbx5. We sought to develop a hypomorphic Zic3 mouse to model human heterotaxy and investigate developmental mechanisms underlying variability in cardiac phenotypes.. Zic3 hypomorphic mice were created by targeted insertion of a neomycin cassette and investigated by gross, histologic, and molecular methods.. Low-level Zic3 expression is sufficient for partial rescue of viability as compared with Zic3 null mice. Concordance of early left-right molecular marker abnormalities and later anatomic abnormalities suggests that the primary effect of Zic3 in heart development occurs during left-right patterning. Cardiac-specific gene expression of Nppa (atrial natriuretic factor) and Tbx5 marked the proper morphological locations in the heart regardless of looping abnormalities.. Zic3 hypomorphic mice are useful models to investigate the variable cardiac defects resulting from a single genetic defect. Low-level Zic3 expression rescues the left pulmonary isomerism identified in Zic3 null embryos. Our data do not support a direct role for Zic3 in the myocardium via regulation of Nppa and Tbx5 and suggest that the primary effect of Zic3 on cardiac development occurs during left-right patterning. Topics: Animals; Atrial Natriuretic Factor; Base Sequence; Disease Models, Animal; DNA Primers; Gene Components; Gene Expression Profiling; Gene Targeting; Heart Defects, Congenital; Heterotaxy Syndrome; Homeodomain Proteins; Humans; Mice; Mice, Mutant Strains; Molecular Sequence Data; Myocardium; Natriuretic Peptide, C-Type; Neomycin; Protein Precursors; Sequence Analysis, DNA; Transcription Factors | 2013 |
C-type natriuretic peptide slows down wound healing but promotes angiogenesis in SKH1-hr hairless mice.
C-type natriuretic peptide (CNP) is known to increase growth rate of endothelial cells in vitro. In addition, gene transfer of CNP into ischaemic muscle was shown to induce angiogenesis. So far, no study has addressed the effect of CNP on dermal wound healing. The ear wound model in mice was used in this study. The first group was treated with dsRed-CNP plasmid, whereas the second group was transfected with the empty dsRed-sine plasmid, lacking sequence coding for CNP. The third group was sham operated and treated with saline to serve as second control. Wound size was measured on days 0, 1, 3, 5, 7, 9, 11 and 14. On days 7 and 14 capillary density was analysed. Wound closure rate was significantly reduced in mice treated with CNP [dsRed-CNP 73·3 ± 3·2% versus dsRed-sine 94·5 ± 2·4% versus saline 92·1 ± 2·4%, n = 8 per group, analysis of variance (ANOVA) P < 0·001] at day 7 postop. Capillary density was found to be significantly higher in CNP-treated mice (dsRed-CNP 18·7 ± 3·9 versus dsRed-sine 12·3 ± 2·7 versus control 10·1 ± 4·7, CD31(+) capillaries per microscope field, ANOVA P = 0·018) at day 14 postoperative. CNP significantly reduces wound closure rate in hairless mice but promotes the development of new blood vessels. A possible explanation is the dual effect of CNP, inhibiting growth of fibromyoblasts but stimulating growth of endothelial cells. Thus, CNP may serve as a therapeutic approach to diseases caused by hyperfibrosis. Topics: Analysis of Variance; Animals; Disease Models, Animal; Endothelium, Vascular; Gene Transfer Techniques; Immunohistochemistry; Male; Mice; Mice, Hairless; Natriuretic Peptide, C-Type; Neovascularization, Physiologic; Random Allocation; Reference Values; Sensitivity and Specificity; Skin; Transfection; Wound Healing; Wounds and Injuries | 2013 |
Pharmacodynamic responses of plasma and tissue C-type natriuretic peptide to GH: correlation with linear growth in GH-deficient rats.
Studies from genetic modification and spontaneous mutations show that C-type natriuretic peptide (CNP) signalling plays an essential part in postnatal endochondral growth, but measurement of CNP proteins and changes in their abundance in tissues and plasma during normal growth has not been reported. Using rodent pups with GH deficiency, we now describe the pharmacodynamic response of CNP and rat amino-terminal proCNP (NTproCNP) in plasma and tissues, and relate these to changes in linear growth (nose-tail length, tibial length and tibial growth plate width) during the course of 1 week of GH or saline (control) administration. Compared with saline, significant increases in plasma and tissue CNP forms were observed after 24 h in GH-treated pups and before any detectable change in linear growth. Whereas CNP abundance was increased in most tissues (muscle, heart and liver) by GH, enrichment was the greatest in extracts from growth plates and kidney. Plasma and tissue concentrations in GH-treated pups were sustained or further increased at 1 week when strong positive associations were found between plasma NTproCNP and linear growth or tissue concentrations. High content of NTproCNP in kidney tissue strongly correlated with plasma concentrations, which is consistent with previous data showing renal extraction of the peptide. In showing a prompt and significant increase in CNP in tissues driving normal endochondral growth, these findings provide further rationale for CNP agonists in the treatment of growth disorders resistant to current therapies and support the use of CNP concentrations as biomarkers of linear growth. Topics: Animals; Biomarkers; Body Weights and Measures; Bone and Bones; Bone Development; Disease Models, Animal; Dwarfism, Pituitary; Growth Hormone; Growth Plate; Hormone Replacement Therapy; Human Growth Hormone; Kidney; Male; Natriuretic Peptide, C-Type; Organ Specificity; Protein Precursors; Rats; Rats, Mutant Strains; Rats, Sprague-Dawley; Recombinant Proteins | 2012 |
Evaluation of the therapeutic potential of a CNP analog in a Fgfr3 mouse model recapitulating achondroplasia.
Achondroplasia (ACH), the most common form of dwarfism, is an inherited autosomal-dominant chondrodysplasia caused by a gain-of-function mutation in fibroblast-growth-factor-receptor 3 (FGFR3). C-type natriuretic peptide (CNP) antagonizes FGFR3 downstream signaling by inhibiting the pathway of mitogen-activated protein kinase (MAPK). Here, we report the pharmacological activity of a 39 amino acid CNP analog (BMN 111) with an extended plasma half-life due to its resistance to neutral-endopeptidase (NEP) digestion. In ACH human growth-plate chondrocytes, we demonstrated a decrease in the phosphorylation of extracellular-signal-regulated kinases 1 and 2, confirming that this CNP analog inhibits fibroblast-growth-factor-mediated MAPK activation. Concomitantly, we analyzed the phenotype of Fgfr3(Y367C/+) mice and showed the presence of ACH-related clinical features in this mouse model. We found that in Fgfr3(Y367C/+) mice, treatment with this CNP analog led to a significant recovery of bone growth. We observed an increase in the axial and appendicular skeleton lengths, and improvements in dwarfism-related clinical features included flattening of the skull, reduced crossbite, straightening of the tibias and femurs, and correction of the growth-plate defect. Thus, our results provide the proof of concept that BMN 111, a NEP-resistant CNP analog, might benefit individuals with ACH and hypochondroplasia. Topics: Achondroplasia; Animals; Bone and Bones; Disease Models, Animal; Growth Plate; Humans; Mice; Mutation; Natriuretic Peptide, C-Type; Organ Size; Radiography; Receptor, Fibroblast Growth Factor, Type 3; Skull; Treatment Outcome | 2012 |
Agents increasing cyclic GMP amplify 5-HT4-elicited positive inotropic response in failing rat cardiac ventricle.
Activation of 5-HT(4) receptors in failing ventricles elicits a cAMP-dependent positive inotropic response which is mainly limited by the cGMP-inhibitable phosphodiesterase (PDE) 3. However, PDE4 plays an additional role which is demasked by PDE3 inhibition. The objective of this study was to evaluate the effect of cGMP generated by particulate and soluble guanylyl cyclase (GC) on the 5-HT(4)-mediated inotropic response. Extensive myocardial infarctions were induced by coronary artery ligation in Wistar rats, exhibiting heart failure 6 weeks after surgery. Contractility was measured in left ventricular preparations. Cyclic GMP was measured by EIA. In ventricular preparations, ANP or BNP displayed no impact on 5-HT(4)-mediated inotropic response. However, CNP increased the 5-HT(4)-mediated inotropic response as well as the β(1)-adrenoceptor (β(1)-AR)-mediated response to a similar extent as PDE3 inhibition by cilostamide. Pretreatment with cilostamide eliminated the effect of CNP. Inhibition of nitric oxide (NO) synthase and soluble GC by L-NAME and ODQ, respectively, attenuated the 5-HT(4)-mediated inotropic response, whereas the NO donor Sin-1 increased this response. The effects were absent during PDE3 inhibition, suggesting cGMP-dependent inhibition of PDE3. However, in contrast to the effects on the 5-HT(4) response, Sin-1 inhibited whereas L-NAME and ODQ enhanced the β(1)-AR-mediated inotropic response. cGMP generated both by particulate (NPR-B) and soluble GC increases the 5-HT(4)-mediated inotropic response in failing hearts, probably through inhibition of PDE3. β(1)-AR and 5-HT(4) receptor signalling are subject to opposite regulatory control by cGMP generated by soluble GC in failing hearts. Thus, cGMP from different sources is functionally compartmented, giving differential regulation of different G(s)-coupled receptors. Topics: Animals; Cyclic GMP; Disease Models, Animal; GTP-Binding Protein alpha Subunits, Gs; Guanylate Cyclase; Heart Failure; Male; Myocardial Contraction; Myocardial Infarction; Natriuretic Peptide, C-Type; Rats; Rats, Wistar; Receptors, Adrenergic, beta-1; Receptors, Atrial Natriuretic Factor; Receptors, Cytoplasmic and Nuclear; Receptors, Serotonin, 5-HT4; Soluble Guanylyl Cyclase | 2011 |
Hypoxia inducible factor-1 improves the negative functional effects of natriuretic peptide and nitric oxide signaling in hypertrophic cardiac myocytes.
Both natriuretic peptides and nitric oxide may be protective in cardiac hypertrophy, although their functional effects are diminished in hypertrophy. Hypoxia inducible factor-1 (HIF-1) may also protect in cardiac hypertrophy. We hypothesized that upregulation of HIF-1 would protect the functional effects of cyclic GMP (cGMP) signaling in hypertrophied ventricular myocytes.. A cardiac hypertrophy model was created in mice by transverse aorta constriction. HIF-1 was increased by deferoxamine (150 mg/kg for 2 days). HIF-1alpha protein levels were examined. Functional parameters were measured (edge detector) on freshly isolated myocytes at baseline and after BNP (brain natriuretic peptide, 10(-8)-10(-7)M) or CNP (C-type natriuretic peptide, 10(-8)-10(-7)M) or SNAP (S-nitroso-N-acetyl-penicillamine, a nitric oxide donor, 10(-6)-10(-5)M) followed by KT5823 (a cyclic GMP-dependent protein kinase (PKG) inhibitor, 10(-6)M). We also determined PKG expression levels and kinase activity.. We found that under control conditions, BNP (-24%), CNP (-22%) and SNAP (-23%) reduced myocyte shortening, while KT5823 partially restored function. Deferoxamine treated control myocytes responded similarly. Baseline function was reduced in the myocytes from hypertrophied heart. BNP, CNP, SNAP and KT5823 also had no significant effects on function in these myocytes. Deferoxamine restored the negative functional effects of BNP (-22%), CNP (-18%) and SNAP (-19%) in hypertrophic cardiac myocytes and KT5823 partially reversed this effect. Additionally, deferoxamine maintained PKG expression levels and activity in hypertrophied heart.. Our results indicated that the HIF-1 protected the functional effects of cGMP signaling in cardiac hypertrophy through preservation of PKG. Topics: Animals; Carbazoles; Cardiomegaly; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Deferoxamine; Disease Models, Animal; Dose-Response Relationship, Drug; Female; Hypoxia-Inducible Factor 1; Hypoxia-Inducible Factor 1, alpha Subunit; Male; Mice; Mice, Inbred C57BL; Myocytes, Cardiac; Natriuretic Peptide, Brain; Natriuretic Peptide, C-Type; Nitric Oxide; S-Nitroso-N-Acetylpenicillamine; Signal Transduction; Up-Regulation | 2010 |
NPR-B, the C-type natriuretic peptide specific receptor, is the predominant biological receptor in mouse and pig myocardial tissue.
The increased myocardial production and the elevated plasma concentrations of C-type natriuretic peptide (CNP) in heart failure patients suggest its involvement in pathophysiological cardiac remodeling. The cardiovascular action of CNP seems to be mainly mediated by natriuretic peptide receptor (NPR)-B but the importance of CNP/NPR-B signaling in heart is not yet well characterized. The aim of this study was to assess the cardiac mRNA expression of CNP and NPR-B together with those of BNP and NPR-A in order to evaluate the relative importance of these peptides and of their receptors in cardiovascular system.. The expression of mRNA coding for CNP, NPR-B, BNP and NPR-A was investigated in myocardial tissue (BALB/c mice, N=5) by use of RT-PCR. NPR-A and NPR-B expression were also evaluated in left ventricle of male adult minipigs without (N=5) and with pacing-induced heart failure (HF, N=5).. The proposed method allowed to detect the expression of mRNA coding for CNP and NPR-B in myocardial tissue confirming the presence of these effectors in the heart. These data also indicate that CNP mRNA expression is lower with respect to that of BNP (CNP/GAPDH= 0.117+/-0.035 vs. BNP/GAPDH=0.247+/-0.066) and that NPR-B is the predominant subtype receptor in the heart (Mouse: NPR-A/GAPDH=0.244+/- 0.028; NPR-B/GAPDH=0.657+/-0.022; p=0.0008; Pig: NPR-A/GAPDH=3.06+/-1.75, NPR-B/GAPDH= 14.3+/-3.6, p=0.0028; HF Pig: NPR-A/GAPDH= 4.29+/-0.93, NPR-B/GAPDH=7.9+/-1.1, p=0.0043).. In the present study, we provided the first evidence of a higher mRNA expression in cardiac tissue of NPR-B with respect to NPR-A indicating that CNP specific receptor (NPR-B) is the predominant biological receptor in mouse and pig myocardial tissue. Topics: Animals; Biomarkers; Disease Models, Animal; Guinea Pigs; Heart Failure; Male; Mice; Mice, Inbred BALB C; Myocardium; Natriuretic Peptide, C-Type; Predictive Value of Tests; Receptors, Atrial Natriuretic Factor; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Sensitivity and Specificity | 2010 |
Distinct regulation of developmental and heart disease-induced atrial natriuretic factor expression by two separate distal sequences.
Nppa, encoding atrial natriuretic factor, is expressed in fetal atrial and ventricular myocardium and is downregulated in the ventricles after birth. During hypertrophy and heart failure, Nppa expression is reactivated in the ventricles and serves as a highly conserved marker of heart disease. The Nppa promoter has become a frequently used model to study mechanisms of cardiac gene regulation. Nevertheless, the regulatory sequences that provide the correct developmental pattern and ventricular reactivation during cardiac disease remain to be defined. We found that proximal Nppa fragments ranging from 250 bp to 16 kbp provide robust reporter gene activity in the atria and correct repression in the atrioventricular canal and the nodes of the conduction system in vivo. However, depending on fragment size and site of integration into the genome of mice, the fetal ventricular activity was either absent or present in an incorrect pattern. Furthermore, these fragments did not provide ventricular reactivation in heart disease models. These results indicate that the proximal promoter does not provide a physiologically relevant model for ventricular gene activity. In contrast, 2 modified bacterial artificial chromosome clones with partially overlapping genomic Nppa sequences provided appropriate reactivation of the green fluorescent protein reporter during pressure overload-induced hypertrophy and heart failure in vivo. However, only 1 of these bacterial artificial chromosomes provided correct fetal ventricular green fluorescent protein activity. These results show that distinct distal regulatory sequences and divergent regulatory pathways control fetal ventricular activity and reactivation of Nppa during cardiac disease, respectively. Topics: Animals; Atrial Natriuretic Factor; Atrioventricular Node; Disease Models, Animal; Gene Expression Regulation, Developmental; Heart Atria; Heart Diseases; Heart Ventricles; Male; Mice; Mice, Transgenic; Natriuretic Peptide, C-Type; Promoter Regions, Genetic; Protein Precursors | 2008 |
Differential interactions of thin filament proteins in two cardiac troponin T mouse models of hypertrophic and dilated cardiomyopathies.
Mutations in a sarcomeric protein can cause hypertrophic cardiomyopathy (HCM) or dilated cardiomyopathy (DCM), the opposite ends of a spectrum of phenotypic responses of the heart to mutations. We posit the contracting phenotypes could result from differential effects of the mutant proteins on interactions among the sarcomeric proteins. To test the hypothesis, we generated transgenic mice expressing either cardiac troponin T (cTnT)-Q92 or cTnT-W141, known to cause HCM and DCM, respectively, in the heart.. We phenotyped the mice by echocardiography, histology and immunoblotting, and real-time polymerase chain reaction. We detected interactions between the sarcomeric proteins by co-immunoprecipitation and determined Ca2+ sensitivity of myofibrillar protein ATPase activity by Carter assay. The cTnT-W141 mice exhibited dilated hearts and decreased systolic function. In contrast, the cTnT-Q92 mice showed smaller ventricles and enhanced systolic function. Levels of cardiac troponin I, cardiac alpha-actin, alpha-tropomyosin, and cardiac troponin C co-immunoprecipitated with anti-cTnT antibodies were higher in the cTnT-W141 than in the cTnT-Q92 mice, as were levels of alpha-tropomyosin co-immunoprecipitated with an anti-cardiac alpha-actin antibody. In contrast, levels of cardiac troponin I co-immunoprecipitated with an anti-cardiac alpha-actin antibody were higher in the cTnT-Q92 mice. Ca2+ sensitivity of myofibrillar ATPase activity was increased in HCM but decreased in DCM mice compared with non-transgenic mice.. Differential interactions among the sarcomeric proteins containing cTnT-Q92 or cTnT-W141 are responsible for the contrasting phenotypes of HCM or DCM, respectively. Topics: Adenosine Triphosphatases; Animals; Atrial Natriuretic Factor; Calcium; Cardiomyopathy, Dilated; Disease Models, Animal; Heart Ventricles; Hypertrophy, Left Ventricular; Mice; Mice, Transgenic; Mutation; Natriuretic Peptide, C-Type; Phenotype; Protein Precursors; Sarcomeres; Troponin T; Ultrasonography | 2008 |
Hyperammonemia inhibits the natriuretic peptide receptor 2 (NPR-2)-mediated cyclic GMP synthesis in the astrocytic compartment of rat cerebral cortex slices.
The decrease of cyclic GMP (cGMP) level in the brain, contributing to cognitive and memory deficit in hyperammonemia (HA), has been attributed to the interference of ammonia with the NMDA/nitric oxide/soluble guanylate cyclase (GC)/cGMP pathway in neurons. The present study tested the hypotheses that (a) HA also affects cGMP synthesis elicited by stimulation of the natriuretic peptide receptor 2 (NPR-2) with its natural ligand, C-type natriuretic peptide (CNP) and (b) the latter effect may involve astrocytes, the ammonia-sensitive cells. In the cerebral cortical slices of control rats, CNP stimulated cGMP synthesis in a degree comparable to the NO donor, S-nitroso-N-acetylpenicillamine (SNAP) used at an optimal concentration. Fluoroacetate (FA), a metabolic inhibitor specifically affecting astrocytic mitochondria, inhibited the CNP-dependent cGMP synthesis by about 50%. Ammonium acetate-induced HA decreased by 68% the CNP-dependent cGMP generation in slices incubated in the absence of FA. In slices incubated in the presence of FA, cGMP synthesis in slices derived from HA rats did not differ from that in control slices. The results indicate that HA inhibits CNP-dependent cGMP synthesis in the FA-vulnerable, astrocytic compartment, but not in the FA-resistant compartment(s) of the brain. HA did not affect the expression of NPR-2 mRNA in the cerebral cortex tissue as tested using real-time PCR, indicating that the effect of ammonia involves as yet unidentified events occurring posttranscriptionally. Deregulation of NPR-2 function in astrocytes by ammonia may contribute to neurophysiological symptoms of HA. Topics: Acetates; Aconitate Hydratase; Animals; Astrocytes; Cerebral Cortex; Cyclic GMP; Disease Models, Animal; Fluoroacetates; Guanylate Cyclase; Hyperammonemia; Male; Natriuretic Peptide, C-Type; Nitric Oxide Donors; Rats; Rats, Wistar; Receptors, Atrial Natriuretic Factor; RNA, Messenger; S-Nitroso-N-Acetylpenicillamine | 2007 |
Negative inotropic effects of C-type natriuretic peptide are attenuated in hypertrophied ventricular myocytes associated with reduced cyclic GMP production.
We tested the hypothesis that the negative inotropic effects of C-type natriuretic peptide (CNP) would be diminished in renal hypertensive (one-kidney-one-clip, 1K1C) hypertrophic rabbit hearts and that this attenuated effect would be due either to decreased cyclic GMP production or to reduced signaling.. Using isolated control and 1K1C ventricular myocytes, cell shortening data (video edge detection) were collected: (1) at baseline and after CNP 10(-8,-7) M, followed by KT5823 (KT), a cyclic GMP-dependent protein kinase inhibitor; or (2) at baseline, following KT pre-treatment and subsequent CNP 10(-8,-7) M. In addition, cyclic GMP levels were determined by radioimmunoassay at baseline and CNP 10(-7) M.. In control myocytes, CNP decreased percent shortening (5.7 +/- 0.4 versus 4.0 +/- 0.4% at 10(-7) M), maximal rate of shortening (58.7 +/- 5.1 versus 45.2 +/- 3.6 microm/sec) and maximal rate of relaxation (57.1 +/- 4.9 versus 44.1 +/- 3.4 microm/sec) in a concentration-dependent manner. These effects were attenuated by subsequent KT administration. CNP failed to produce these negative functional effects in 1K1C myocytes. When pre-treated with KT, CNP had no negative functional effect in either normal and 1K1C myocytes. Basal levels of cyclic GMP were similar in control versus 1K1C myocytes; however, CNP produced a significant rise in cyclic GMP level in control (63.6 +/- 7.8 versus 83.5 +/- 11.3 pmol/10(5) myocytes) but not in 1K1C (49.2 +/- 2.6 versus 52.7 +/- 5.6) myocytes.. Thus, CNP acted through the cyclic GMP protein kinase in control myocytes. We conclude that in hypertrophic cardiac myocytes, the decreased effect of CNP was because of decreased production of cyclic GMP. Topics: Animals; Carbazoles; Cardiomegaly; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Disease Models, Animal; Guanylate Cyclase; Heart Ventricles; Hypertension, Renal; Hypertrophy; Indoles; Myocardial Contraction; Myocytes, Cardiac; Natriuretic Peptide, C-Type; Protein Kinase Inhibitors; Rabbits; Signal Transduction; Surgical Instruments | 2006 |
Functional effects of C-type natriuretic peptide and nitric oxide are attenuated in hypertrophic myocytes from pressure-overloaded mouse hearts.
Increases in the myocardial level of cGMP usually exert negative inotropic effects in the mammalian hearts. We tested the hypothesis that the negative functional effects caused by nitric oxide (NO) or C-type natriuretic peptide (CNP) through cGMP would be blunted in hypertrophied cardiac myocytes. Contractile function, guanylyl cyclase activity, cGMP-dependent protein phosphorylation, and calcium transients were assessed in ventricular myocytes from aortic stenosis-induced hypertrophic and age-matched control mice. Basal percentage shortening was similar in control and hypertrophic myocytes. S-nitroso-N-acetyl-penicillamine (SNAP, an NO donor, 10(-6) and 10(-5) M) or CNP (10(-8) and 10(-7) M) reduced percentage shortening in both groups, but their effects were blunted in hypertrophic myocytes. Maximal rates of shortening and relaxation were depressed at the basal level, and both reagents had attenuated effects in hypertrophy. Similar results were also found after treatment with guanylin and carbon monoxide, other stimulators of particulate, and soluble guanylyl cyclase, respectively. Guanylyl cyclase activity was not significantly changed in hypertrophy. Addition of Rp-8-[(4-chlorophenyl)thio]-cGMPS triethylamine (an inhibitor of cGMP-dependent protein kinase, 5 x 10(-6) M) blocked SNAP or the effect of CNP in control mice but not in hypertrophy, indicating the cGMP-dependent kinase (PKG) may not mediate the actions of cGMP induced by NO or CNP in the hypertrophic state. Calcium transients after SNAP or CNP were not significantly changed in hypertrophy. These results suggest that in hypertrophied mice, diminished effects of NO or CNP on ventricular myocyte contraction are not due to changes in guanylyl cyclase activity. The data also indicated that PKG-mediated pathways were diminished in hypertrophied myocardium, contributing to blunted effects. Topics: Animals; Aortic Valve Stenosis; Blood Pressure; Cardiomegaly; Cyclic GMP; Disease Models, Animal; Female; Male; Mice; Mice, Inbred C57BL; Myocardial Contraction; Myocytes, Cardiac; Natriuretic Peptide, C-Type; Nitric Oxide; Organ Size | 2005 |
C-type natriuretic peptide inhibits constrictive remodeling without compromising re-endothelialization in balloon-dilated renal arteries.
To investigate the long-term effect of local, liposome-mediated gene transfer of C-type natriuretic peptide (CNP) plasmid versus CNP protein on restenosis in porcine renal arteries following balloon angioplasty.. The renal arteries of 15 pigs were dilated and the adventitia at the site of balloon injury injected with CNP protein, pCR3.1 plasmid encoding CNP, or the beta-galactosidase gene (control) via a needle injection catheter. Five animals receiving the CNP and control genes in dilated arteries were sacrificed after 3 weeks to analyze re-endothelialization, proliferation, and early CNP expression. Ten animals designated for the long-term experiments (3 months) were treated with the CNP gene versus CNP protein (n=3), the CNP gene versus the control gene (n=3), and the CNP protein versus the control gene (n=3). One animal served as a dilated non-treated control. Transfection and expression of CNP and beta-galactosidase were measured by polymerase chain reaction (PCR) and reverse transcription PCR. Renal arterial lumen narrowing was measured with angiography and histology. Endothelialization was assessed using Evans blue stain; vWF, CD31, factor VIII, and Ki67 were markers for immunohistochemical analysis.. An intact endothelial layer was seen at 3 weeks following angioplasty in all transfected arteries. Three months following treatment, computer-assisted morphometric analysis revealed significant enlargement of the arterial cross-sectional areas in CNP plasmid- treated vessels compared to dilated but untreated arteries (CNP plasmid +34.8%+/-13.9% versus CNP protein -1.75%+/-19.9% versus beta-galactosidase -47.0%+/-13.9%, p<0.01). Angiographic analysis showed significant enlargement of the arterial diameter compared to dilated, untreated arteries (CNP plasmid +20.8%+/-6.8% versus CNP protein +5.7%+/-6.0% versus beta-galactosidase -24.5%+/-10.2%, p<0.01).. Local application of CNP plasmid proved superior to CNP protein in producing rapid re-endothelialization and significantly enlarging the renal arterial lumen following dilation. Topics: Angioplasty, Balloon; Animals; Disease Models, Animal; Endothelium, Vascular; Liposomes; Natriuretic Agents; Natriuretic Peptide, C-Type; Plasmids; Regeneration; Renal Artery Obstruction; Secondary Prevention; Swine; Time Factors; Transfection | 2005 |
CNP gene expression is activated by Wnt signaling and correlates with Wnt4 expression during renal injury.
C-type natriuretic peptide (CNP) regulates salt excretion, vascular tone, and fibroblast proliferation and activation. CNP inhibits fibroblast activation in vitro and fibrosis in vivo, but endogenous CNP gene (Nppc) expression during tissue fibrosis has not been reported. We determined that Nppc is induced in renal tubular epithelia and then in interstitial myofibroblasts after unilateral ureteral obstruction (UUO). Induction of Nppc occurred in identical cell populations to those in which Wnt4 is induced after renal injury. In addition, Nppc was activated in Wnt4-expressing cells during nephrogenesis. Wnt signaling components beta-catenin and T cell factor/lymphoid enhancer binding factor (TCF/LEF) specifically bound to cognate elements in the Nppc proximal promoter. Wnt-4, beta-catenin, and LEF-1 activated an Nppc transgene in cultured cells, and transgene activation by Wnt-4 and LEF-1 was dependent on the presence of intact cognate elements. These findings suggest that Wnt-4 stimulates Nppc in a TCF/LEF-dependent manner after renal injury and thus may contribute to limiting renal fibrosis. Topics: Animals; Cell Line; Disease Models, Animal; DNA-Binding Proteins; Gene Expression Regulation, Developmental; Kidney; Kidney Tubules; Lymphoid Enhancer-Binding Factor 1; Mice; Natriuretic Peptide, C-Type; Proto-Oncogene Proteins; RNA, Messenger; Signal Transduction; Transcription Factors; Ureteral Obstruction; Wnt Proteins; Wnt4 Protein; Zebrafish Proteins | 2003 |
Local expression of C-type natriuretic peptide suppresses inflammation, eliminates shear stress-induced thrombosis, and prevents neointima formation through enhanced nitric oxide production in rabbit injured carotid arteries.
We previously observed that adenovirus-mediated expression of C-type natriuretic peptide (CNP) markedly inhibits neointima formation after balloon injury in rat carotid arteries, suggesting that CNP has multiple effects over its modest inhibitory effect on cellular proliferation. We hypothesized that local expression of CNP might have antithrombotic and antiinflammatory effects. Balloon-injured rabbit carotid arteries were infected with an adenovirus expressing human CNP (AdCNP), human tissue factor pathway inhibitor (AdTFPI), or bacterial beta-galactosidase (AdLacZ) or infused with saline. Seven days later, shear stress-induced thrombosis was evaluated by cyclic flow variation (CFV), reflecting recurrent cycles of thrombus formation and dislodgment. CFV was observed in all AdLacZ-infected and saline-infused arteries but not in arteries infected with AdCNP or AdTFPI even in the presence of epinephrine. Injury increased the expressions of intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and infiltration of macrophages. However, these effects were markedly reduced in AdCNP-treated arteries but not in AdTFPI-infected ones. In AdCNP-infected arteries, injury-induced expression of inducible NO synthase (iNOS) was enhanced, leading to increased NO generation. Interestingly, when the enhanced NO production was inhibited, neither inhibitory effect was observed, and suppression of neointima formation by CNP was canceled. Our study demonstrates that overexpression of CNP shows antithrombotic and antiinflammatory effects and reduces neointima formation mainly through enhanced NO production. Topics: Adenoviridae; Animals; beta-Galactosidase; Blood Flow Velocity; Carotid Arteries; Carotid Artery Thrombosis; Disease Models, Animal; Enzyme Inhibitors; Genes, Reporter; Genetic Therapy; Genetic Vectors; Humans; Inflammation; Intercellular Adhesion Molecule-1; Lipoproteins; Macrophages; Male; Natriuretic Peptide, C-Type; Nitric Oxide; Rabbits; Rats; Rats, Wistar; Stress, Mechanical; Tunica Intima; Vascular Cell Adhesion Molecule-1; Vasoconstrictor Agents | 2002 |
Dwarfism and early death in mice lacking C-type natriuretic peptide.
Longitudinal bone growth is determined by endochondral ossification that occurs as chondrocytes in the cartilaginous growth plate undergo proliferation, hypertrophy, cell death, and osteoblastic replacement. The natriuretic peptide family consists of three structurally related endogenous ligands, atrial, brain, and C-type natriuretic peptides (ANP, BNP, and CNP), and is thought to be involved in a variety of homeostatic processes. To investigate the physiological significance of CNP in vivo, we generated mice with targeted disruption of CNP (Nppc(-/-) mice). The Nppc(-/-) mice show severe dwarfism as a result of impaired endochondral ossification. They are all viable perinatally, but less than half can survive during postnatal development. The skeletal phenotypes are histologically similar to those seen in patients with achondroplasia, the most common genetic form of human dwarfism. Targeted expression of CNP in the growth plate chondrocytes can rescue the skeletal defect of Nppc(-/-) mice and allow their prolonged survival. This study demonstrates that CNP acts locally as a positive regulator of endochondral ossification in vivo and suggests its pathophysiological and therapeutic implication in some forms of skeletal dysplasia. Topics: Animals; Bone and Bones; Chondrocytes; Disease Models, Animal; Dwarfism; Female; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Natriuretic Peptide, C-Type; Ossification, Heterotopic | 2001 |
Compared effects of natriuretic peptides on ovalbumin-induced asthmatic model.
We compared the effects of natriuretic peptides on antigen-induced bronchoconstriction and airway microvascular leakage in sensitized guinea pigs. Anesthetized male guinea pigs, ventilated via a tracheal cannula, were placed in a plethysmograph to measure pulmonary mechanics for 10 min after challenge with 1 mg/kg of ovalbumin, and then Evans blue dye was extravasated into airway tissue in order to indicate and evaluate microvascular leakage. Three separate intravenous pretreatments using atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP) significantly inhibited the ovalbumin-induced bronchoconstriction and microvascular leakage in a dose-dependent manner. These inhibitory effects were mimicked by 8-bromoguanosine 3',5'-cyclic monophosphate. We showed that the rank order of inhibitory potencies, which were mediated by cyclic guanosine 3',5'-monophosphate, was BNP > or = ANP > or = CNP. These results gave us some clues for the clinical application of the natriuretic peptides. Topics: Animals; Antigens; Asthma; Atrial Natriuretic Factor; Blood Pressure; Bronchi; Bronchoconstriction; Capillary Permeability; Cyclic GMP; Disease Models, Animal; Guinea Pigs; Leukotriene D4; Male; Natriuretic Peptide, Brain; Natriuretic Peptide, C-Type; Nerve Tissue Proteins; Ovalbumin; Proteins; Trachea | 1998 |
Neutral endopeptidase inhibition potentiates the effects of natriuretic peptides in renin transgenic rats.
The influence of neutral endopeptidase (NEP) inhibition with (S)-thiorphan on the hormonal, renal, and blood-pressure-lowering effects of an infusion of atrial (ANP), brain (BNP), and C-type natriuretic peptide (CNP) was evaluated in hypertensive transgenic rats (TGR) harboring an additional mouse renin gene (TGR(m(Ren2)27)). These TGR possess an activated natriuretic peptide system as compared with Sprague-Dawley rats (SDR), used in this study as control. (S)-Thiorphan significantly decreased blood pressure in anesthetized TGR but not in anesthetized SDR during the 60-min infusion period. Exogenously administered ANP decreased blood pressure in SDR with no significant effects in TGR after 60 min. In contrast, BNP infusion significantly decreased blood pressure in TGR, while changes in SDR were not significant. The blood pressure was further decreased after combined infusion of ANP and BNP with (S)-thiorphan in TGR. No effect on blood pressure was registered during infusion of CNP in either experimental group. The plasma levels of ANP, BNP, and cGMP were higher in TGR than in SDR, whereas plasma renin activity was lower. Co-administration of ANP, BNP, or CNP with the NEP inhibitor (S)-thiorphan potentiated the plasma ANP, BNP, and cGMP. Infusion of ANP alone did not affect BNP plasma levels of TGR and vice versa. In contrast, CNP infusion increased ANP plasma levels in both TGR and SDR. Renal excretion of sodium and cGMP increased after infusion of (S)-thiorphan and ANP or BNP in both TGR and SDR. The combination of ANP and (S)-thiorphan had a slightly greater effect on urinary excretion of sodium and cGMP in TGR than either compound alone, but the effects were more pronounced in SDR than in TGR. Finally, infusion of CNP alone and in combination with (S)-thiorphan influenced the excretion of sodium and cyclic GMP only slightly. These results indicate that inhibition of neutral endopeptidase by (S)-thiorphan potentiates the hemodynamic and renal effects of natriuretic peptides ANP and BNP, and to some extent those of CNP, in hypertensive TGR and normotensive SDR. In contrast to ANP and BNP, infusion of CNP had no effect on the blood pressure in anesthetized TGR or SDR. Inhibition of NEP therefore seems to be a promising way to potentiate endogenous levels of natriuretic peptides, which may be of therapeutic benefit in cardiovascular diseases such as hypertension or heart failure. Topics: Animals; Animals, Genetically Modified; Atrial Natriuretic Factor; Blood Pressure; Cyclic GMP; Disease Models, Animal; Drug Therapy, Combination; Electrolytes; Hypertension; Mice; Natriuretic Peptide, Brain; Natriuretic Peptide, C-Type; Neprilysin; Protease Inhibitors; Proteins; Rats; Rats, Sprague-Dawley; Renin; Sodium; Thiorphan | 1996 |